Developing therapeutic approaches for metachromatic leukodystrophy

Shilpa A Patil, Gustavo H B Maegawa, Shilpa A Patil, Gustavo H B Maegawa

Abstract

Metachromatic leukodystrophy (MLD) is an autosomal recessive lysosomal disorder caused by the deficiency of arylsulfatase A (ASA), resulting in impaired degradation of sulfatide, an essential sphingolipid of myelin. The clinical manifestations of MLD are characterized by progressive demyelination and subsequent neurological symptoms resulting in severe debilitation. The availability of therapeutic options for treating MLD is limited but expanding with a number of early stage clinical trials already in progress. In the development of therapeutic approaches for MLD, scientists have been facing a number of challenges including blood-brain barrier (BBB) penetration, safety issues concerning therapies targeting the central nervous system, uncertainty regarding the ideal timing for intervention in the disease course, and the lack of more in-depth understanding of the molecular pathogenesis of MLD. Here, we discuss the current status of the different approaches to developing therapies for MLD. Hematopoietic stem cell transplantation has been used to treat MLD patients, utilizing both umbilical cord blood and bone marrow sources. Intrathecal enzyme replacement therapy and gene therapies, administered locally into the brain or by generating genetically modified hematopoietic stem cells, are emerging as novel strategies. In pre-clinical studies, different cell delivery systems including microencapsulated cells or selectively neural cells have shown encouraging results. Small molecules that are more likely to cross the BBB can be used as enzyme enhancers of diverse ASA mutants, either as pharmacological chaperones, or proteostasis regulators. Specific small molecules may also be used to reduce the biosynthesis of sulfatides, or target different affected downstream pathways secondary to the primary ASA deficiency. Given the progressive neurodegenerative aspects of MLD, also seen in other lysosomal diseases, current and future therapeutic strategies will be complementary, whether used in combination or separately at specific stages of the disease course, to produce better outcomes for patients afflicted with this devastating inherited disorder.

Keywords: arylsulfatase A; enzyme enhancement therapy; enzyme replacement therapy; gene therapy; metachromatic leukodystrophy; small molecules.

Figures

Figure 1
Figure 1
Sulfatide metabolic pathway. Notes: The figure shows the biosynthetic pathways of sulfatide, the sphingolipid that accumulates during metachromatic leukodystrophy MLD. The specific enzymes described in each step of the pathway are in italics. The rounded boxes depict the products or substrates of the reactions. The green shaded square shapes show different therapeutic approaches for MLD treatment. Two current substrate reduction therapies for another lysosomal disease (Gaucher disease) are also shown: N-Butyldeoxynojirimycin (NB-DNJ) miglustat (Zavesca®, Actelion Pharmaceuticals Ltd, Allschwil, Switzerland), and eliglustat tartrate. Both of these small molecules competitively inhibit ceramide glucosyltransferase. Abbreviations: ASA, arylsulfatase A; EET, enzyme enhancement therapy; ERT, enzyme replacement therapy; Gal, galactose; HSCT, hematopoietic stem cell transplantation; Glc, glucose; SGal-Glc-cer, sulfo-galactose-glucosylceramide; MLD, metachromatic leukodystrophy; SRT, substrate reduction therapy.
Figure 2
Figure 2
Schematic representation of the mechanism of action of enzyme enhancement agents including pharmacological chaperones (PCs) and proteostasis regulators (PRs). Notes: ASA is synthesized in the endoplasmic reticulum (ER) as several other proteins target to different intracellular organelles, plasma membrane, and those destined to be secreted to the extracellular environment. Concomitant to the translation process, nascent ASA peptide interacts with several components of the ER folding system. Once folded properly, ASA is targeted to the ER exit pathway and ultimately reaches the lysosomal compartment, where it exerts its physiological function. Missense mutations in the ARSA gene can affect physicochemical interactions between amino acids, resulting in impairment of the folding process of the mutant ASA. PCs (blue boxes) are small molecules that physically interact with the target protein, in this case ASA, and assist its folding, preventing it from being targeted to the ERAD pathway and ultimately degraded by the ubiquitin-proteasomal system. Thus, increased levels of mutant ASA reach the lysosomes to degrade the natural substrate (sulfatides). eventually, other small molecules function as PRs (beige/brown boxes), which can improve the cellular folding capacity by interacting with ER folding components. These molecules can also promote physiological cellular responses, such as unfolded protein response, which can be advantageous to enhance folding of specific misfolded ASA mutants. Other PRs (red boxes) can also potentially inhibit components of ERAD and favor the interactions of misfolded ASA mutants with the ER folding elements, increasing the probability of reaching a folding-like conformation. Abbreviations: ASA, arylsulfatase A; ER, endoplasmic reticulum; ERAD, endoplasmic reticulum associate degradation; PC, pharmacological chaperones; PR, proteostasis regulators.
Figure 3
Figure 3
Cell-based high-throughput screening (HTS) assay. Notes: Here, a cell-based HTS assay for galactocerebrosidase (GALC), which is the lysosomal enzyme deficient in Krabbe disease or globoid cell leukodystrophy (GLD), is depicted for (A and B). The 1,536-well plate contains GLD patient cells that are cultured and treated with a small molecule collection for a specific period of time. After the treatment period, the specific enzyme GALC can be measured using a fluorescent substrate, 6-hexadecanoylamino-4-methylumbelliferyl-β-D-galactoside (HMU) for the enzyme. In each 1,536-well plate, control cells (columns 2 and 3) are seeded and later assayed. These control cells have normal Gal-ase enzymatic activity and are not treated with the library. The columns 1 and 2 contain only culture medium and no cells, and are used as backgrounds for the assays. Columns 5–48 contain cultured patients cells (B)– in this case, fibroblasts. Each well (yellow square) corresponds to a specific small molecule from the library, which is tested in different concentrations. (C) Two-plate view diagrams are shown with the fluorescent signals generated from the HMU against the different rows of a 1,536-well plate. Substantial assay window is observed between GALC enzymatic activity of controls (triangles) and GLD patient cells (circles). The small molecule candidates are selected based on the enhancement of the residual GALC enzyme activity. (D) The quantitative nature of the HTS assay, where seven different concentrations of the target library were tested, allows the generation of concentration-response curves (CRCs) given by the HMU fluorescence signal increase. Based on the characteristics of the CRCs, prioritization of “hits” can be done during the validation process with secondary and tertiary assays.

References

    1. Austin J, McAfee D, Armstrong D, O’Rourke M, Shearer L, Bachhawat B. Low sulfatase activities in metachromatic leukodystrophy (MLD). A controlled study of enzymes in 9 living and 4 autopsied patients with MLD. Trans Am Neurol Assoc. 1964;89:147–150.
    1. Bosio A, Binczek E, Stoffel W. Functional breakdown of the lipid bilayer of the myelin membrane in central and peripheral nervous system by disrupted galactocerebroside synthesis. Proc Natl Acad Sci U S A. 1996;1293(23):13280–13285.
    1. Aggarwal S, Yurlova L, Simons M. Central nervous system myelin: structure, synthesis and assembly. Trends Cell Biol. 2011;21(10):585–593.
    1. Faust PL, Kaye EM, Powers JM. Myelin lesions associated with lysosomal and peroxisomal disorders. Expert Rev Neurother. 2010;10(9):1449–1466.
    1. Gieselmann V. Metachromatic leukodystrophy: genetics, pathogenesis and therapeutic options. Acta Paediatr Suppl. 2008;97(457):15–21.
    1. Mechtler TP, Stary S, Metz TF, et al. Neonatal screening for lysosomal storage disorders: feasibility and incidence from a nationwide study in Austria. Lancet. 2012;379(9813):335–341.
    1. Meikle PJ, Hopwood JJ, Clague AE, Carey WF. Prevalence of lysosomal storage disorders. JAMA. Jan. 1999;281(3):249–254.
    1. Von Figura K. Metachromatic Leukodystrophy. In: Scriver C, Beaudet A, Arthur L, Sly WS, Valle D, editors. Metabolic and Molecular Basis of Inherited Disease. Vol. 3. New York: McGraw-Hill; 2001. pp. 3696–3724.
    1. Lugowska A, Poninska J, Krajewski P, Broda G, Ploski R. Population carrier rates of pathogenic ARSA gene mutations: is metachromatic leukodystrophy underdiagnosed? PLoS One. 2011;6(6):e20218.
    1. Stenson PD, Ball EV, Howells K, Phillips AD, Mort M, Cooper DN. The Human Gene Mutation Database: providing a comprehensive central mutation database for molecular diagnostics and personalized genomics. Hum Genomics. 2009;4(2):69–72.
    1. Gieselmann V. An assay for the rapid detection of the arylsulfatase A pseudodeficiency allele facilitates diagnosis and genetic counseling for metachromatic leukodystrophy. Hum Genet. 1991;86(3):251–255.
    1. Gieselmann V, Polten A, Kreysing J, von Figura K. Molecular genetics of metachromatic leukodystrophy. J Inherit Metab Dis. 1994;17(4):500–509.
    1. Gort L, Coll MJ, Chabas A. Identification of 12 novel mutations and two new polymorphisms in the arylsulfatase A gene: haplotype and genotype-phenotype correlation studies in Spanish metachromatic leukodystrophy patients. Hum Mutat. 1999;14(3):240–248.
    1. Francis GS, Bonni A, Shen N, et al. Metachromatic leukodystrophy: multiple nonfunctional and pseudodeficiency alleles in a pedigree: problems with diagnosis and counseling. Ann Neurol. 1993;34(2):212–218.
    1. Hohenschutz C, Eich P, Friedl W, Waheed A, Conzelmann E, Propping P. Pseudodeficiency of arylsulfatase A: a common genetic polymorphism with possible disease implications. Hum Genet. 1989;82(1):45–48.
    1. Nelson PV, Carey WF, Morris CP. Population frequency of the arylsulphatase A pseudo-deficiency allele. Hum Genet. 19987(1):87–88.
    1. Ott R, Waye JS, Chang PL. Evolutionary origins of two tightly linked mutations in arylsulfatase-A pseudodeficiency. Hum Genet. 1997;101(2):135–140.
    1. Sommerlade HJ, Hille-Rehfeld A, von Figura K, Gieselmann V. Four monoclonal antibodies inhibit the recognition of arylsulphatase A by the lysosomal enzyme phosphotransferase. Biochem J. 1994;297(Pt 1):123–130.
    1. Fluharty AL, Fluharty CB, Bohne W, von Figura K, Gieselmann V. Two new arylsulfatase A (ARSA) mutations in a juvenile metachromatic leukodystrophy (MLD) patient. Am J Hum Genet. 1991;49(6):1340–1350.
    1. Harvey JS, Nelson PV, Carey WF, Robertson EF, Morris CP. An arylsulfatase A (ARSA) missense mutation (T274M) causing late-infantile metachromatic leukodystrophy. Hum Mutat. 1993;2(4):261–267.
    1. Ghosh P, Dahms NM, Kornfeld S. Mannose 6-phosphate receptors: new twists in the tale. Nat Rev Mol Cell Biol. 2003;4(3):202–212.
    1. Lukatela G, Krauss N, Theis K, et al. Crystal structure of human arylsulfatase A: the aldehyde function and the metal ion at the active site suggest a novel mechanism for sulfate ester hydrolysis. Biochemistry. 1998;37(11):3654–3664.
    1. Von Bulow R, Schmidt B, Dierks T, et al. Defective oligomerization of arylsulfatase a as a cause of its instability in lysosomes and metachromatic leukodystrophy. J Biol Chem. 2002;277(11):9455–9461.
    1. von Bulow R, Schmidt B, Dierks T, von Figura K, Uson I. Crystal structure of an enzyme-substrate complex provides insight into the interaction between human arylsulfatase A and its substrates during catalysis. J Mol Biol. 2001;305(2):269–277.
    1. Siegel EG, Lucke H, Schauer W, Creutzfeldt W. Repeated upper gastrointestinal hemorrhage caused by metachromatic leukodystrophy of the gall bladder. Digestion. 1992;51(2):121–124.
    1. Ries M, Deeg KH. Polyposis of the gallbladder associated with metachromatic leukodystrophy. Eur J Pediatr. 1993;152(5):450–451.
    1. Kim TS, Kim IO, Kim WS, Choi YS, Yeon KM, Hwang YS. Involvement of the gallbladder in childhood metachromatic leukodystrophy: ultrasonographic findings. J Ultrasound Med. 1996;15(12):821–825.
    1. Oak S, Rao S, Karmarkar S, et al. Papillomatosis of the gallbladder in metachromatic leukodystrophy. Pediatr Surg Int. 1997;12(5–6):424–425.
    1. Simanovsky N, Ackerman Z, Kiderman A, Fields S. Unusual gallbladder findings in two brothers with metachromatic leukodystrophy. Pediatr Radiol. 1998;28(9):706–708.
    1. Krivan HC, Olson LD, Barile MF, Ginsburg V, Roberts DD. Adhesion of Mycoplasma pneumoniae to sulfated glycolipids and inhibition by dextran sulfate. J Biol Chem. 1989;264(16):9283–9288.
    1. Sugano K, Tai T, Kawashima I, et al. Localization of sulfatides in the epithelial lining of gastric mucosa: studies with a monoclonal antibody to sulfatides. J Clin Gastroenterol. 1995;21(Suppl 1):S98–S103.
    1. Takamatsu K. Phytosphingosine-containing neutral glycosphingolipids and sulfatides in the human female genital tract: their association in the cervical epithelium and the uterine endometrium and their dissociation in the mucosa of fallopian tube with the menstrual cycle. Keio J Med. 1992;41(3):161–167.
    1. Malone MJ, Stoffyn P. A comparative study of brain and kidney glycolipids in metachromatic leucodystrophy. J Neurochem. 1966;13(11):1037–1045.
    1. Toda K, Kobayashi T, Goto I, Kurokawa T, Ogomori K. Accumulation of lysosulfatide (sulfogalactosylsphingosine) in tissues of a boy with metachromatic leukodystrophy. Biochem Biophys Res Commun. 1989;15159(2):605–611.
    1. Toda K, Kobayashi T, Goto I, et al. Lysosulfatide (sulfogalactosylsphingosine) accumulation in tissues from patients with metachromatic leukodystrophy. J Neurochem. 1990;55(5):1585–1591.
    1. Natowicz MR, Prence EM, Chaturvedi P, Newburg DS. Urine sulfatides and the diagnosis of metachromatic leukodystrophy. Clin Chem. 1996;2(2):232–238.
    1. Honke K, Zhang Y, Cheng X, Kotani N, Taniguchi N. Biological roles of sulfoglycolipids and pathophysiology of their deficiency. Glycoconj J. 2004;21(1–2):59–62.
    1. Vitner EB, Platt FM, Futerman AH. Common and uncommon pathogenic cascades in lysosomal storage diseases. J Biol Chem. 2010;285(27):20423–20427.
    1. Webster HD. Schwann cell alterations in metachromatic leukodystrophy: preliminary phase and electron microscopic observations. J Neuropathol Exp Neurol. 1962;21:534–554.
    1. Peng L, Suzuki K. Ultrastructural study of neurons in metachromatic leukodystrophy. Clin Neuropathol. 1987;6(5):224–230.
    1. Berger J, Loschl B, Bernheimer H, et al. Occurrence, distribution, and phenotype of arylsulfatase A mutations in patients with metachromatic leukodystrophy. Am J Med Genet. 1997;69(3):335–340.
    1. Polten A, Fluharty AL, Fluharty CB, Kappler J, von Figura K, Gieselmann V. Molecular basis of different forms of metachromatic leukodystrophy. N Engl J Med. 1991;24(1):18–22.
    1. Gieselmann V, Krageloh-Mann I. Metachromatic leukodystrophy – an update. Neuropediatrics. 2010;41(1):1–6.
    1. Poeppel P, Habetha M, Marcao A, Bussow H, Berna L, Gieselmann V. Missense mutations as a cause of metachromatic leukodystrophy. Degradation of arylsulfatase A in the endoplasmic reticulum. FEBS J. 2005;272(5):1179–1188.
    1. Poeppel P, Abouzied MM, Volker C, Gieselmann V. Misfolded endoplasmic reticulum retained subunits cause degradation of wild-type subunits of arylsulfatase A heteromers. FEBS J. 2010;277(16):3404–3414.
    1. Hagberg B. Clinical aspects of globoid cell and metachromatic leukodystrophies. Birth Defects Orig Art Ser. Feb. 1971;7(1):103–112.
    1. Hess MW, Kirschning E, Pfaller K, Debbage PL, Hohenberg H, Klima G. 5000-year-old myelin: uniquely intact in molecular configuration and fine structure. Curr Biol. 1998;8(15):R512–R513.
    1. Zoller I, Meixner M, Hartmann D, et al. Absence of 2-hydroxylated sphingolipids is compatible with normal neural development but causes late-onset axon and myelin sheath degeneration. J Neurosci. 2008;2428(39):9741–9754.
    1. O’Brien JS, Sampson EL. Myelin Membrane: a molecular abnormality. Science. 1965;150(3703):1613–1614.
    1. Tanaka K, Nagara H, Kobayashi T, Goto I. The twitcher mouse: accumulation of galactosylsphingosine and pathology of the central nervous system. Brain Res. 1989;482(2):347–350.
    1. Coetzee T, Li X, Fujita N, et al. Molecular cloning, chromosomal mapping, and characterization of the mouse UDP-galactose:ceramide galactosyltransferase gene. Genomics. 1996;35(1):215–222.
    1. Coetzee T, Fujita N, Dupree J, et al. Myelination in the absence of galactocerebroside and sulfatide: normal structure with abnormal function and regional instability. Cell. 1996;86(2):209–219.
    1. Coetzee T, Suzuki K, Popko B. New perspectives on the function of myelin galactolipids. Trends Neurosci. 1998;21(3):126–130.
    1. Meixner M, Jungnickel J, Grothe C, Gieselmann V, Eckhardt M. Myelination in the absence of UDP-galactose:ceramide galactosyltransferase and fatty acid 2 -hydroxylase. BMC Neurosci. 2011;12:22.
    1. Potter KA, Kern MJ, Fullbright G, et al. Central nervous system dysfunction in a mouse model of FA2H deficiency. Glia. 2011;59(7):1009–1021.
    1. Imgrund S, Hartmann D, Farwanah H, et al. Adult ceramide synthase 2 (CERS2)-deficient mice exhibit myelin sheath defects, cerebellar degeneration, and hepatocarcinomas. J Biol Chem. 2009;284(48):33549–33560.
    1. Pewzner-Jung Y, Park H, Laviad EL, et al. A critical role for ceramide synthase 2 in liver homeostasis: I. alterations in lipid metabolic pathways. The Journal of biological chemistry. 2010;285(14):10902–10910.
    1. von Figura K. Metachromatic Leukodystrophy. In: Scriver C, Beaudet A, Arthur L, Sly WS, Valle D, editors. Metabolic and Molecular Basis of Inherited Disease. Vol. 3. New York: McGrw-Hill; 2001. pp. 3696–3724.
    1. Hess B, Saftig P, Hartmann D, et al. Phenotype of arylsulfatase A-deficient mice: relationship to human metachromatic leukodystrophy. Proc Natl Acad Sci U S A. 1996;93(25):14821–14826.
    1. Eckhardt M, Hedayati KK, Pitsch J, et al. Sulfatide storage in neurons causes hyperexcitability and axonal degeneration in a mouse model of metachromatic leukodystrophy. J Neurosci. 2007;27(34):9009–9021.
    1. Walkley SU. Pathogenic mechanisms in lysosomal disease: a reappraisal of the role of the lysosome. Acta Paediatr Suppl. 2007;96(455):26–32.
    1. Walkley SU. Pathogenic cascades in lysosomal disease-Why so complex? J Inherit Metab Dis. 2009;32(2):181–189.
    1. Paez PM, Fulton D, Colwell CS, Campagnoni AT. Voltage-operated Ca(2+) and Na(+) channels in the oligodendrocyte lineage. J Neurosci Res. 2009;87(15):3259–3266.
    1. Paez PM, Fulton DJ, Spreuer V, et al. Golli myelin basic proteins regulate oligodendroglial progenitor cell migration through voltagegated Ca2+ influx. J Neurosci. 2009;29(20):6663–6676.
    1. Pelled D, Trajkovic-Bodennec S, Lloyd-Evans E, Sidransky E, Schiffmann R, Futerman AH. Enhanced calcium release in the acute neuronopathic form of Gaucher disease. Neurobiol Dis. 2005;18(1):83–88.
    1. Ausseil J, Desmaris N, Bigou S, et al. Early neurodegeneration progresses independently of microglial activation by heparan sulfate in the brain of mucopolysaccharidosis IIIB mice. PLoS One. 2008;3(5):e2296.
    1. Shimada Y, Kobayashi H, Kawagoe S, et al. Endoplasmic reticulum stress induces autophagy through activation of p38 MAPK in fibroblasts from Pompe disease patients carrying c.546G>T mutation. Mol Genet Metab. 2011;104(4):566–573.
    1. Warr MR, Binnewies M, Flach J, et al. FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature. 2013;494(7437):323–327.
    1. Nishiyama Y, Shimada Y, Yokoi T, et al. Akt inactivation induces endoplasmic reticulum stress-independent autophagy in fibroblasts from patients with Pompe disease. Mol Genet Metab. 2012;107(3):490–495.
    1. Lloyd-Evans E, Morgan AJ, He X, et al. Niemann-Pick disease type C1 is a sphingosine storage disease that causes deregulation of lysosomal calcium. Nat Med. 2008;14(11):1247–1255.
    1. Chen CS, Patterson MC, Wheatley CL, O’Brien JF, Pagano RE. Broad screening test for sphingolipid-storage diseases. Lancet. 1999;354(9182):901–905.
    1. Mukherjee S, Soe TT, Maxfield FR. Endocytic sorting of lipid analogues differing solely in the chemistry of their hydrophobic tails. J Cell Biol. 1999;144(6):1271–1284.
    1. Settembre C, Fraldi A, Jahreiss L, et al. A block of autophagy in lysosomal storage disorders. Hum Mol Genet. 2008;17(1):119–129.
    1. Settembre C, Arteaga-Solis E, McKee MD, et al. Proteoglycan desulfation determines the efficiency of chondrocyte autophagy and the extent of FGF signaling during endochondral ossification. Genes Dev. 2008;22(19):2645–2650.
    1. Settembre C, Annunziata I, Spampanato C, et al. Systemic inflammation and neurodegeneration in a mouse model of multiple sulfatase deficiency. Proc Natl Acad Sci U S A. 2007;104(11):4506–4511.
    1. Dierks T, Schmidt B, Borissenko LV, et al. Multiple sulfatase deficiency is caused by mutations in the gene encoding the human C(alpha)-formylglycine generating enzyme. Cell. 2003;113(4):435–444.
    1. Lo EH. Degeneration and repair in central nervous system disease. Nat Med. 2010;16(11):1205–1209.
    1. Pardridge WM. The blood-brain barrier: bottleneck in brain drug development. NeuroRx. 2005;2(1):3–14.
    1. Ghose AK, Viswanadhan VN, Wendoloski JJ. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J Comb Chem. 1999;1(1):55–68.
    1. Priller J, Flugel A, Wehner T, et al. Targeting gene-modified hematopoietic cells to the central nervous system: use of green fluorescent protein uncovers microglial engraftment. Nat Med. 2001;7(12):1356–1361.
    1. Mehl E, Jatzkewitz H. A cerebrosidesulfatase from swine kidney. Hoppe Seylers Z Physiol Chem. 1964;339(1):260–276.
    1. Saravanan K, Schaeren-Wiemers N, Klein D, et al. Specific downregulation and mistargeting of the lipid raft-associated protein MAL in a glycolipid storage disorder. Neurobiol Dis. 2004;16(2):396–406.
    1. Wittke D, Hartmann D, Gieselmann V, Lullmann-Rauch R. Lysosomal sulfatide storage in the brain of arylsulfatase A-deficient mice: cellular alterations and topographic distribution. Acta Neuropathol. 2004;108(4):261–271.
    1. Krivit W, Shapiro E, Kennedy W, et al. Treatment of late infantile metachromatic leukodystrophy by bone marrow transplantation. N Engl J Med. 1990;322(1):28–32.
    1. Martin HR, Poe MD, Provenzale JM, Kurtzberg J, Mendizabal A, Escolar ML. Neurodevelopmental outcomes of umbilical cord blood transplantation in metachromatic leukodystrophy. Biol Blood Marrow Transplant. 2013;19(4):616–624.
    1. Malm G, Ringden O, Winiarski J, et al. Clinical outcome in four children with metachromatic leukodystrophy treated by bone marrow transplantation. Bone Marrow Transplant. 1996;17(6):1003–1008.
    1. Nagai T, Kaneko T, Shichijou H, Karato T, Maruyama A, Tsuchiya Y. Bone marrow transplantation for late infantile metachromatic leukodystrophy: pathogenic investigation for graft rejection. Acta Paediatr Jpn. 1993;35(5):404–408.
    1. Navarro C, Dominguez C, Fernandez JM, Fachal C, Alvarez M. Case report: four-year follow-up of bone-marrow transplantation in late juvenile metachromatic leukodystrophy. J Inherit Metab Dis. 1995;18(2):157–158.
    1. Solders G, Celsing G, Hagenfeldt L, Ljungman P, Isberg B, Ringden O. Improved peripheral nerve conduction, EEG and verbal IQ after bone marrow transplantation for adult metachromatic leukodystrophy. Bone Marrow Transplant. 1998;22(11):1119–1122.
    1. Walkley SU, Wurzelmann S, Siegel DA. Ectopic axon hillockassociated neurite growth is maintained in metabolically reversed swainsonine-induced neuronal storage disease. Brain Res. 1987;410(1):89–96.
    1. Matzner U, Lullmann-Rauch R, Stroobants S, et al. Enzyme replacement improves ataxic gait and central nervous system histopathology in a mouse model of metachromatic leukodystrophy. Mol Ther. 2009;17(4):600–606.
    1. Matthes F, Stroobants S, Gerlach D, et al. Efficacy of enzyme replacement therapy in an aggravated mouse model of metachromatic leukodystrophy declines with age. Hum Mol Genet. 2012;21(11):2599–2609.
    1. Martino S, Consiglio A, Cavalieri C, et al. Expression and purification of a human, soluble Arylsulfatase A for Metachromatic Leukodystrophy enzyme replacement therapy. J Biotechnol. 2005;117(3):243–251.
    1. Asheuer M, Pflumio F, Benhamida S, et al. Human CD34+ cells differentiate into microglia and express recombinant therapeutic protein. Proc Nat Acad Sci U S A. 2004;101(10):3557–3562.
    1. Saftig P, Klumperman J. Lysosome biogenesis and lysosomal membrane proteins: trafficking meets function. Nat Rev Mol Cell Biol. 2009;10(9):623–635.
    1. Bredius RG, Laan LA, Lankester AC, et al. Early marrow transplantation in a pre-symptomatic neonate with late infantile metachromatic leukodystrophy does not halt disease progression. Bone Marrow Transplant. 2007;39(5):309–310.
    1. Krivit W, Peters C, Shapiro EG. Bone marrow transplantation as effective treatment of central nervous system disease in globoid cell leukodystrophy, metachromatic leukodystrophy, adrenoleukodystrophy, mannosidosis, fucosidosis, aspartylglucosaminuria, Hurler, Maroteaux-Lamy, and Sly syndromes, and Gaucher disease type III. Curr Opin Neurol. 1999;12(2):167–176.
    1. Malatack JJ, Consolini DM, Bayever E. The status of hematopoietic stem cell transplantation in lysosomal storage disease. Pediatr Neurol. 2003;29(5):391–403.
    1. Peters C, Steward CG. Hematopoietic cell transplantation for inherited metabolic diseases: an overview of outcomes and practice guidelines. Bone Marrow Transplant. 2003;31(4):229–239.
    1. Biffi A, Lucchini G, Rovelli A, Sessa M. Metachromatic leukodystrophy: an overview of current and prospective treatments. Bone Marrow Transplant. 2008;42(Suppl 2):S2–S6.
    1. Orchard PJ, Tolar J. Transplant outcomes in leukodystrophies. Semin Hematol. 2010;47(1):70–78.
    1. Batzios S P, Zafeiriou DI. Developing treatment options for metachromatic leukodystrophy. Mol Genet Metab. 2012;105(1):56–63.
    1. Matzner U, Gieselmann V. Gene therapy of metachromatic leukodystrophy. Expert Opin Biol Ther. 2005;5(1):55–65.
    1. Biffi A, De Palma M, Quattrini A, et al. Correction of metachromatic leukodystrophy in the mouse model by transplantation of genetically modified hematopoietic stem cells. J Clin Invest. 2004;113(8):1118–1129.
    1. Biffi A, Capotondo A, Fasano S, et al. Gene therapy of metachromatic leukodystrophy reverses neurological damage and deficits in mice. J Clin Invest. 2006;116(11):3070–3082.
    1. Matzner U, Hartmann D, Lullmann-Rauch R, et al. Bone marrow stem cell-based gene transfer in a mouse model for metachromatic leukodystrophy: effects on visceral and nervous system disease manifestations. Gene Ther. 2002;9(1):53–63.
    1. Capotondo A, Cesani M, Pepe S, et al. Safety of arylsulfatase A overexpression for gene therapy of metachromatic leukodystrophy. Hum Gene Ther. 2007;18(9):821–836.
    1. Consiglio A, Martino S, Dolcetta D, et al. Metabolic correction in oligodendrocytes derived from metachromatic leukodystrophy mouse model by using encapsulated recombinant myoblasts. J Neurol Sci. 2007;255(1–2):7–16.
    1. Lagranha VL, Baldo G, de Carvalho TG, et al. In vitro correction of ARSA deficiency in human skin fibroblasts from metachromatic leukodystrophy patients after treatment with microencapsulated recombinant cells. Metab Brain Dis. 2008;23(4):469–484.
    1. Givogri MI, Galbiati F, Fasano S, et al. Oligodendroglial progenitor cell therapy limits central neurological deficits in mice with metachromatic leukodystrophy. J Neurosci. 2006;26(12):3109–3119.
    1. Kawabata K, Migita M, Mochizuki H, et al. Ex vivo cell-mediated gene therapy for metachromatic leukodystrophy using neurospheres. Brain Res. 2006;1094(1):13–23.
    1. Givogri MI, Bottai D, Zhu HL, et al. Multipotential neural precursors transplanted into the metachromatic leukodystrophy brain fail to generate oligodendrocytes but contribute to limit brain dysfunction. Dev Neurosci. 2008;30(5):340–357.
    1. Klein D, Schmandt T, Muth-Kohne E, et al. Embryonic stem cell-based reduction of central nervous system sulfatide storage in an animal model of metachromatic leukodystrophy. Gene Ther. 2006;13(24):1686–1695.
    1. Zervas M, Somers KL, Thrall MA, Walkley SU. Critical role for glycosphingolipids in Niemann-Pick disease type C. Curr Biol. 20011(16):1283–1287.
    1. Platt FM, Neises GR, Dwek RA, Butters TD. N-butyldeoxynojirimycin is a novel inhibitor of glycolipid biosynthesis. J Biol Chem. 1994;269(11):8362–8365.
    1. Grabowski GA, Osiecki-Newman K, Dinur T, et al. Human acid betaglucosidase. Use of conduritol B epoxide derivatives to investigate the catalytically active normal and Gaucher disease enzymes. J Biol Chem. 1986;261(18):8263–8269.
    1. Adachi M, Volk BW. Gaucher disease in mice induced by conduritol-B-epoxide: morphologic features. Arch Pathol Lab Med. 1977;101(5):255–259.
    1. Platt FM, Jeyakumar M. Substrate reduction therapy. Acta Paediatr Suppl. 2008;97(457):88–93.
    1. Platt FM, Neises GR, Reinkensmeier G, et al. Prevention of lysosomal storage in Tay-Sachs mice treated with N-butyldeoxynojirimycin. Science. 1997;276(5311):428–431.
    1. Maegawa GH, van Giersbergen PL, Yang S, et al. Pharmacokinetics, safety and tolerability of miglustat in the treatment of pediatric patients with GM2 gangliosidosis. Mol Genet Metab. 2009;97(4):284–291.
    1. Maegawa GH, Banwell BL, Blaser S, et al. Substrate reduction therapy in juvenile GM2 gangliosidosis. Mol Genet Metab. 2009;98(1–2):215–224.
    1. Patterson MC, Vecchio D, Prady H, Abel L, Wraith JE. Miglustat for treatment of Niemann-Pick C disease: a randomised controlled study. Lancet Neurol. 2007;6(9):765–772.
    1. Sundaram KS, Lev M. Warfarin administration reduces synthesis of sulfatides and other sphingolipids in mouse brain. J Lipid Res. 1988 Nov;29(11):1475–1479.
    1. Assadi M, Wang DJ, Anderson K, Carran M, Bilaniuk L, Leone P. Vitamin k antagonist warfarin for palliative treatment of metachromatic leukodystrophy, a compassionate study of four subjects. J Cent Nerv Syst Dis. 2012;4:73–79.
    1. Bernier V, Lagace M, Bichet DG, Bouvier M. Pharmacological chaperones: potential treatment for conformational diseases. Trends Endocrinol Metab. 2004;15(5):222–228.
    1. Leinekugel P, Michel S, Conzelmann E, Sandhoff K. Quantitative correlation between the residual activity of beta-hexosaminidase A and arylsulfatase A and the severity of the resulting lysosomal storage disease. Hum Genet. 1992;88(5):513–523.
    1. Wiseman RL, Powers ET, Buxbaum JN, Kelly JW, Balch WE. An adaptable standard for protein export from the endoplasmic reticulum. Cell. 2007;131(4):809–821.
    1. Maegawa GH, Tropak M, Buttner J, et al. Pyrimethamine as a potential pharmacological chaperone for late-onset forms of GM2 gangliosidosis. J Biol Chem. 2007;282(12):9150–9161.
    1. Maegawa GH, Tropak MB, Buttner JD, et al. Identification and characterization of ambroxol as an enzyme enhancement agent for Gaucher disease. J Biol Chem. 2009;284(35):23502–23516.
    1. Beck M. Emerging drugs for lysosomal storage diseases. Expert Opin Emerg Drugs. 2010;15(3):495–507.
    1. Fan JQ. A counterintuitive approach to treat enzyme deficiencies: use of enzyme inhibitors for restoring mutant enzyme activity. Biol Chem. 2008;389(1):1–11.
    1. Balch WE, Morimoto RI, Dillin A, Kelly JW. Adapting proteostasis for disease intervention. Science. 2008;319(5865):916–919.
    1. Harbut MB, Patel BA, Yeung BK, et al. Targeting the ERAD pathway via inhibition of signal peptide peptidase for antiparasitic therapeutic design. Proc Natl Acad Sci U S A. 2012;109(52):21486–21491.
    1. Elfrink HL, Zwart R, Baas F, Scheper W. Inhibition of endoplasmic reticulum associated degradation reduces endoplasmic reticulum stress and alters lysosomal morphology and distribution. Mol Cells. 2013;35(4):291–297.
    1. Wang F, Segatori L. Remodeling the proteostasis network to rescue glucocerebrosidase variants by inhibiting ER-associated degradation and enhancing ER folding. PLoS One. 2013;8(4):e61418.
    1. Mu TW, Ong DS, Wang YJ, et al. Chemical and biological approaches synergize to ameliorate protein-folding diseases. Cell. 2008;134(5):769–781.
    1. Mu TW, Fowler DM, Kelly JW. Partial restoration of mutant enzyme homeostasis in three distinct lysosomal storage disease cell lines by altering calcium homeostasis. PLoS Biol. 2008;6(2):e26.
    1. Yang C, Rahimpour S, Lu J, et al. Histone deacetylase inhibitors increase glucocerebrosidase activity in Gaucher disease by modulation of molecular chaperones. Proc Natl Acad Sci U S A. 2013;110(3):966–971.
    1. An WF, Tolliday N. Cell-based assays for high-throughput screening. Mol Biotechnol. 2010;45(2):180–186.
    1. Hampton SL, Kinnaird AI. Genetic interventions in mammalian cells; applications and uses in high-throughput screening and drug discovery. Cell Biol Toxicol. 2010;26(1):43–55.
    1. Geng H, Whiteley G, Ribbens J, et al. Novel patient cell-based HTS assay for identification of small molecules for a lysosomal storage disease. PLoS One. 2011;6(12):e29504.
    1. Ribbens J, Whiteley G, Furuya H, et al. A high-throughput screening assay using Krabbe disease patient cells. Anal Biochem. 2012;434(1):15–25.
    1. Pelled D, Lloyd-Evans E, Riebeling C, Jeyakumar M, Platt FM, Futerman AH. Inhibition of calcium uptake via the sarco/endoplasmic reticulum Ca2+-ATPase in a mouse model of Sandhoff disease and prevention by treatment with N-butyldeoxynojirimycin. J Biol Chem. 2003;278(32):29496–29501.
    1. Jeyakumar M, Smith DA, Williams IM, et al. NSAIDs increase survival in the Sandhoff disease mouse: synergy with N-butyldeoxynojirimycin. Ann Neurol. 2004;56(5):642–649.
    1. Smith D, Wallom K-L, Williams IM, Jeyakumar M, Platt FM. Beneficial effects of anti-inflammatory therapy in a mouse model of Niemann-Pick disease type C1. Neurobiol Dis. 2009;36(2):242–251.
    1. Inglese J, Auld DS, Jadhav A, et al. Quantitative high-throughput screening: a titration-based approach that efficiently identifies biological activities in large chemical libraries. Proc Natl Acad Sci U S A. 2006;103(31):11473–11478.
    1. National Center for Research Resources (NCRR) Stem cell transplantation (SCT) for genetic diseases Available from . NLM identifier NCT00004378Accessed July 8, 2013
    1. Masonic Cancer Center, University of Minnesota Stem cell transplant for inborn errors of metabolism Available from . /NLM identifier NCT00176904Accessed July 8, 2013
    1. Shire Human Genetic Therapies, Inc Long-term Metazym treatment of patients with late infantile metachromatic leukodystrophy (MLD) Available from . NLM identifier NCT00633139Accessed July 8, 2013
    1. Shire Human Genetic Therapies, Inc Multicenter study of HGT-1110 enzyme replacement therapy administered intrathecally in children with metachromatic leukodystrophy (IDEAMLD) Available from . NLM identifier NCT0150039Accessed July 8, 2013
    1. Assistance Publique – Hôpitaux de Paris Efficacy of Metazym for the treatment of metachromatic leukodystrophy treated with hematopoietic stem cell transplantation (Azylis) Available at . NLM identifier NCT 01303146Accessed July 8, 2013
    1. Shire Human Genetic Therapies, Inc Open-label extension study of recombinant human arylsulfatase A (HGT-1111) in late infantile MLD Available at . NLM identifier NCT 00681811Accessed July 22, 2013
    1. Shire Human Genetic Therapies, Inc Metazym for the treatment of patients with late infantile metachromatic leukodystrophy Available from . NLM identifier NCT00418561Accessed July 8, 2013
    1. IRCCS San Raffaele Gene therapy for metachromatic leukodystrophy (TIGET-MLD Available from . NLM identifier NCT01560182Accessed July 8, 2013
    1. Institut National de la Santé Et de la Recherche Médicale, France Intracerebral gene therapy for children with early onset forms of metachromatic leukodystrophy (TG-MLD) Available from . NLM identifier NCT 01801709Accessed July 8, 2013
    1. The Cooper Health System Effect of warfarin in the treatment of metachromatic leukodystrophy Available at . NLM identifer NCT00683189Accessed July 8, 2013

Source: PubMed

3
Abonnere